Review
Mitophagy and Alzheimer’s Disease: Cellular and Molecular Mechanisms

https://doi.org/10.1016/j.tins.2017.01.002Get rights and content

Trends

Mitochondrial homeostasis is important for synaptic plasticity, learning, and memory.

Neurons affected in Alzheimer’s disease (AD) exhibit dysfunctional mitochondria.

Mitophagy plays important roles in mitochondrial homeostasis, neuroprotection, and resistance to neurodegeneration.

Induced pluripotent stem cell-derived neurons and animal models of sporadic AD provide powerful tools to investigate mitochondrial dysfunction and mitophagy.

NAD+ depletion and impaired mitophagy may occur early in AD and contribute to synaptic dysfunction and neuronal degeneration.

Neurons affected in Alzheimer’s disease (AD) experience mitochondrial dysfunction and a bioenergetic deficit that occurs early and promotes the disease-defining amyloid beta peptide (Aβ) and Tau pathologies. Emerging findings suggest that the autophagy/lysosome pathway that removes damaged mitochondria (mitophagy) is also compromised in AD, resulting in the accumulation of dysfunctional mitochondria. Results in animal and cellular models of AD and in patients with sporadic late-onset AD suggest that impaired mitophagy contributes to synaptic dysfunction and cognitive deficits by triggering Aβ and Tau accumulation through increases in oxidative damage and cellular energy deficits; these, in turn, impair mitophagy. Interventions that bolster mitochondrial health and/or stimulate mitophagy may therefore forestall the neurodegenerative process in AD.

Section snippets

Early Clues That Mitochondria Are Central to AD

AD is the most common form of dementia and is characterized by a progression from episodic memory problems to severe cognitive decline and complete dependence of the patient on caregivers 1, 2, 3. The disease-defining histopathological abnormalities – extracellular deposits of Aβ and intraneuronal accumulation of hyperphosphorylated Tau (pTau) – ‘spread’ through the brain in a nonrandom manner with early pathology occurring in the entorhinal cortex and hippocampus 4, 5. However, the alterations

Mitochondria and Neuroplasticity

Mitochondria are organelles referred to as the ‘powerhouses’ of cells. Along with regulating calcium homeostasis and signaling to and from other organelles, mitochondria produce ATP by oxidative phosphorylation, in which electrons are passed through the electron transport chain from high-energy substrates to oxygen. Impaired mitochondrial function may lead to a reduction in cellular energy levels; concomitant leakage of electrons promotes the formation of reactive oxygen species (ROS), which

Mitophagy: Molecular Machinery and Regulatory Mechanisms

Autophagy is an evolutionarily conserved process in which cytoplasmic substrates are engulfed in autophagic vesicles, fused to lysosomes, and degraded. Autophagy is classified into various subgroups based on the mechanism of substrate delivery to the lysosome; these groups are macroautophagy, chaperone-mediated autophagy, and microautophagy 18, 20. We focus on macroautophagy (hereafter refer to as autophagy), in which engulfment by a double-membrane autophagosomal structure is followed by

Mitochondrial Dysfunction in AD

Studies in living AD patients and postmortem brain tissue have provided evidence that neurons in affected brain regions suffer impaired mitochondrial function. PET brain scans reveal decreased radiolabeled glucose uptake into neurons and biochemical analyses demonstrate reduced activity of mitochondrial enzymes involved in oxidative phosphorylation and the TCA cycle [6]. Recent findings suggest that mitochondrial biogenesis is impaired in AD as indicated by reduced levels of the transcriptional

Compromised Autophagy and Mitophagy in AD

Increasing evidence suggests that inhibition of the clearance of damaged mitochondria, along with concurrent increases in oxidative stress levels, results in the accumulation of dysfunctional neurons in AD. To be removed by mitophagy, the autophagosome containing the mitochondrion must fuse with a lysosome to form an autolysosome in which proteases degrade the mitochondrion (Figure 1A). Neurons exhibiting abnormal accumulation of autophagosomal vacuoles are a prominent feature in AD and their

Using iPSCs to Study Mitochondrial Function and Mitophagy in AD

The mitochondrial etiology of AD discussed here suggests that animal or cellular models with familial AD-associated genetic mutations, while of significant use, may not fully recapitulate important aspects of AD onset and progression. Studies using cells and tissue from human AD patients may better illustrate the nature and extent of mitophagy and mitochondrial dysfunction and iPSCs represent a useful tool for such studies. Since the discovery of iPSCs in 2006 by Yamanaka and colleagues [97],

Stimulating Mitophagy As an Approach to Delaying and Treating AD

Pharmacological agents and lifestyle interventions aimed at improving mitochondrial health and enhancing mitophagy have been evaluated in animal models and, in some cases, in mild cognitive impairment (MCI) or AD patients (Figure 3). Caloric restriction, intermitting fasting, and vigorous exercise are bioenergetic challenges that can promote neuroplasticity (synapse formation, hippocampal neurogenesis, and learning and memory) and bolster neuronal stress resistance 111, 112. Evidence from

Concluding Remarks

Significant progress has been made in ascertaining the causes of the devastating neurodegenerative disorder AD. The accumulation of dysfunctional mitochondria has emerged as a common feature of affected neurons in patients and animal models that may occur before discernible cognitive deficits. Experimental manipulations that impair mitophagy can enhance Aβ and pTau pathologies, while interventions that stimulate mitophagy can preserve synaptic plasticity and cognitive function. In addition,

Acknowledgments

The authors acknowledge the valuable work of the many investigators whose published articles they were unable to cite owing to space limitations. They thank Drs Emmette Hutchison and Krisztina Marosi for critical reading of the manuscript, Dr Nuo Sun for his comments and inputs, and Marc Raley for generation of the figures. This research was supported by the Intramural Research Program of the National Institute on Aging, including two NIA intralaboratory grants (2015-2016, 2016-2017 to

References (123)

  • D.C. Rubinsztein

    Autophagy and aging

    Cell

    (2011)
  • A. Rakovic

    Phosphatase and tensin homolog (PTEN)-induced putative kinase 1 (PINK1)-dependent ubiquitination of endogenous Parkin attenuates mitophagy: study in human primary fibroblasts and induced pluripotent stem cell-derived neurons

    J. Biol. Chem.

    (2013)
  • M.P. Mattson

    Energy intake and exercise as determinants of brain health and vulnerability to injury and disease

    Cell Metab.

    (2012)
  • D. Cao

    Intake of sucrose-sweetened water induces insulin resistance and exacerbates memory deficits and amyloidosis in a transgenic mouse model of Alzheimer disease

    J. Biol. Chem.

    (2007)
  • V.K. Halagappa

    Intermittent fasting and caloric restriction ameliorate age-related behavioral deficits in the triple-transgenic mouse model of Alzheimer’s disease

    Neurobiol. Dis.

    (2007)
  • M.J. Schafer

    Reduction of β-amyloid and γ-secretase by calorie restriction in female Tg2576 mice

    Neurobiol. Aging

    (2015)
  • L. Chen

    NLRP3 inflammasome activation by mitochondrial reactive oxygen species plays a key role in long-term cognitive impairment induced by paraquat exposure

    Neurobiol. Aging

    (2015)
  • L. Buee

    Tau protein isoforms, phosphorylation and role in neurodegenerative disorders

    Brain Res. Brain Res. Rev.

    (2000)
  • Y. Hou

    Permeability transition pore-mediated mitochondrial superoxide flashes mediate an early inhibitory effect of amyloid β1-42 on neural progenitor cell proliferation

    Neurobiol. Aging

    (2014)
  • D.C. David

    Proteomic and functional analyses reveal a mitochondrial dysfunction in P301L Tau transgenic mice

    J. Biol. Chem.

    (2005)
  • D. Liu

    Nicotinamide forestalls pathology and cognitive decline in Alzheimer mice: evidence for improved neuronal bioenergetics and autophagy procession

    Neurobiol. Aging

    (2013)
  • J.H. Lee

    Lysosomal proteolysis and autophagy require presenilin 1 and are disrupted by Alzheimer-related PS1 mutations

    Cell

    (2010)
  • E.E. Coffey

    Lysosomal alkalization and dysfunction in human fibroblasts with the Alzheimer’s disease-linked presenilin 1 A246E mutation can be reversed with cAMP

    Neuroscience

    (2014)
  • A.M. Labrousse

    C. elegans dynamin-related protein DRP-1 controls severing of the mitochondrial outer membrane

    Mol. Cell

    (1999)
  • B. DuBoff

    Why size matters – balancing mitochondrial dynamics in Alzheimer’s disease

    Trends Neurosci.

    (2013)
  • X. Wang

    Dynamin-like protein 1 reduction underlies mitochondrial morphology and distribution abnormalities in fibroblasts from sporadic Alzheimer’s disease patients

    Am. J. Pathol.

    (2008)
  • E.F. Fang

    NAD+ replenishment improves lifespan and healthspan in ataxia telangiectasia models via mitophagy and DNA repair

    Cell Metabolism.

    (2016)
  • E.F. Fang

    Defective mitophagy in XPA via PARP-1 hyperactivation and NAD+/SIRT1 reduction

    Cell

    (2014)
  • A.H. Tseng

    SIRT3 deacetylates FOXO3 to protect mitochondria against oxidative damage

    Free Radic. Biol. Med.

    (2013)
  • S. Martire

    PARP-1 involvement in neurodegeneration: a focus on Alzheimer’s and Parkinson’s diseases

    Mech. Ageing Dev.

    (2015)
  • K. Takahashi et al.

    Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors

    Cell

    (2006)
  • M. Citron

    Alzheimer’s disease: strategies for disease modification

    Nat. Rev. Drug Discov.

    (2010)
  • J.P. Blass et al.

    The role of oxidative abnormalities in the pathophysiology of Alzheimer’s disease

    Rev. Neurol. (Paris)

    (1991)
  • D.C. Wallace

    Mitochondrial diseases in man and mouse

    Science

    (1999)
  • A. Cheng

    Involvement of PGC-1α in the formation and maintenance of neuronal dendritic spines

    Nat. Commun.

    (2012)
  • F.M. Menzies

    Compromised autophagy and neurodegenerative diseases

    Nat. Rev. Neurosci.

    (2015)
  • Q. Cai et al.

    Alterations in mitochondrial quality control in Alzheimer’s disease

    Front. Cell. Neurosci.

    (2016)
  • E.F. Fang

    Nuclear DNA damage signalling to mitochondria in ageing

    Nat. Rev. Mol. Cell Biol.

    (2016)
  • K. Palikaras

    Coordination of mitophagy and mitochondrial biogenesis during ageing in C. elegans

    Nature

    (2015)
  • R.J. Youle et al.

    Mechanisms of mitophagy

    Nat. Rev. Mol. Cell Biol.

    (2011)
  • M. Lazarou

    The ubiquitin kinase PINK1 recruits autophagy receptors to induce mitophagy

    Nature

    (2015)
  • S. Geisler

    PINK1/Parkin-mediated mitophagy is dependent on VDAC1 and p62/SQSTM1

    Nat. Cell Biol.

    (2010)
  • F.H. Sterky

    Impaired mitochondrial transport and Parkin-independent degeneration of respiratory chain-deficient dopamine neurons in vivo

    Proc. Natl. Acad. Sci. U. S. A.

    (2011)
  • F. Strappazzon

    AMBRA1 is able to induce mitophagy via LC3 binding, regardless of PARKIN and p62/SQSTM1

    Cell Death Differ.

    (2015)
  • M. Chen

    Mitophagy receptor FUNDC1 regulates mitochondrial dynamics and mitophagy

    Autophagy

    (2016)
  • H. Sandoval

    Essential role for Nix in autophagic maturation of erythroid cells

    Nature

    (2008)
  • F. Gao

    The mitochondrial protein BNIP3L is the substrate of PARK2 and mediates mitophagy in PINK1/PARK2 pathway

    Hum. Mol. Genet.

    (2015)
  • P. Spilman

    Inhibition of mTOR by rapamycin abolishes cognitive deficits and reduces amyloid-β levels in a mouse model of Alzheimer’s disease

    PLoS One

    (2010)
  • J.G. Geisler

    DNP, mitochondrial uncoupling, and neuroprotection: a little dab’ll do ya

    Alzheimers Dement.

    (2016)
  • A.M. Stranahan

    Diet-induced insulin resistance impairs hippocampal synaptic plasticity and cognition in middle-aged rats

    Hippocampus

    (2008)
  • Cited by (0)

    View full text